1
|
Fujimoto A, Koutake Y, Tsutsui Y, Nakahara M, Matsuo K, Yabuuchi Y, Kamimura G, Kawamata Y, Uehara T, Ikari A, Endo S, Oyamada J. Effect of antiemetic corticosteroids on the development of immune-related adverse events caused by chemoimmunotherapy: a multicenter retrospective study. Support Care Cancer 2025; 33:204. [PMID: 39971819 DOI: 10.1007/s00520-025-09268-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE Chemoimmunotherapy is the primary treatment approach for non-small cell lung cancer (NSCLC); however, it is associated with immune-related adverse events (irAEs). Corticosteroids can control irAEs through their anti-inflammatory and immunosuppressive effects. Dexamethasone (DEX) is a potent corticosteroid commonly used to prevent chemotherapy-induced nausea and vomiting (CINV). This study aimed to the association of corticosteroids used to alleviate CINV and irAE occurrence. METHODS This retrospective study included patients with NSCLC who underwent chemoimmunotherapy across eight hospitals. Cases lacking aprepitant use were excluded. All corticosteroids for CINV were standardized to intravenous DEX doses, and cutoff values were calculated using receiver operating characteristic curve analysis. Logistic regression analysis was performed to investigate irAE risk factors. RESULTS The cutoff value for DEX was 15.9 mg (area under the curve, 0.58; 95% confidence interval, 0.45-0.70; sensitivity, 0.63; specificity, 0.61), with 99 and 76 patients in the DEX < 15.9 and ≥ 15.9 mg groups, respectively. Patients in the DEX < 15.9 mg group had a significantly higher incidence of irAE than patients in the DEX ≥ 15.9 mg group (P = 0.03). Multivariate analysis identified that DEX < 15.9 mg was a risk factor for irAEs (P = 0.04; odds ratio: 2.51; 95% confidence interval, 1.03-6.09). CONCLUSION Corticosteroids with DEX equivalent doses of < 15.9 mg in combination with aprepitant for CINV may elevate the risk of irAEs. Therefore, diligent monitoring for irAEs occurrence is warranted in regimens utilizing DEX-equivalent corticosteroid doses of < 15.9 mg combined with aprepitant for CINV prevention.
Collapse
Affiliation(s)
- Airi Fujimoto
- Department of Pharmacy, NHO Beppu Medical Center, 1473 Uchikamado, Beppu, Oita, 874-0011, Japan.
| | - Yoshimichi Koutake
- Department of Pharmacy, Clinical Research Institute, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Yuki Tsutsui
- Department of Pharmacy, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Moeko Nakahara
- Department of Pharmacy, Clinical Research Institute, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Keisuke Matsuo
- Department of Pharmacy, NHO Miyakonojo Medical Center, Miyazaki, Japan
| | - Yurika Yabuuchi
- Department of Pharmacy, NHO Fukuokahigashi Medical Center, Fukuoka, Japan
| | - Go Kamimura
- Department of Pharmacy, NHO Minamikyusyu Hospital, Kagoshima, Japan
| | - Yosei Kawamata
- Department of Pharmacy, NHO Miyazaki Higashi Hospital, Miyazaki, Japan
| | - Tomohiro Uehara
- Department of Pharmacy, NHO Okinawa National Hospital, Okinawa, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, Japan
| | - Satoshi Endo
- The United Graduate School of and Medical Information Sciences, Gifu University, Gifu, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| | - Junji Oyamada
- Department of Pharmacy, NHO Beppu Medical Center, 1473 Uchikamado, Beppu, Oita, 874-0011, Japan
| |
Collapse
|
2
|
Ji X, Huang Z, Zhou C, Wang Y, Geng D, Zhang G, Kang Y, Cui R, Wang J, Zhang T. Esketamine alleviates depressive-like behavior in neuropathic pain mice through the METTL3-GluA1 pathway. Cell Biol Toxicol 2025; 41:38. [PMID: 39875576 PMCID: PMC11775062 DOI: 10.1007/s10565-024-09975-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/21/2024] [Indexed: 01/30/2025]
Abstract
Esketamine, a newly developed antidepressant, is the subject of this research which seeks to explore its impact on depressive symptoms in neuropathic pain mice and the potential molecular mechanisms involved. Through transcriptome sequencing and bioinformatics analysis combined with in vivo studies, it was identified that esketamine markedly boosts the levels of the m6A methyltransferase METTL3 and the AMPA receptor GluA1 subunit. Esketamine activates METTL3, allowing it to bind with GluA1 mRNA, promoting m6A modification, thereby enhancing GluA1 expression at synapses. Through this mechanism, esketamine may reduce depressive-like behavior in neuropathic pain mice, providing new insights into the potential applications of esketamine and novel therapeutic avenues for neuropathic pain and depressive behavior.
Collapse
Affiliation(s)
- Xiaoming Ji
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhimin Huang
- Department of Stomatology, Peoples Hospital, Shizhu Tujia Autonomous County, Chongqing, 409100, China
| | - Chenming Zhou
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu Wang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Dongliang Geng
- Changan Dental Clinic, Xinqidian Dental Medical Services Co., Ltd, Shijiazhuang, 050000, China
| | - Guoliang Zhang
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yunxiao Kang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Rui Cui
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jinyang Wang
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Tianyun Zhang
- Laboratory of Neurobiology, Hebei Medical University, Shijiazhuang, 050017, China.
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Center for Brain Science and Disease, Hebei Medical University, Shijiazhuang, 050017, China.
- Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, 050017, China.
- Laboratory of Neurobiology, Hebei Medical University, Zhongshan Donglu No.361, Shijiazhuang, 050017, China.
| |
Collapse
|
3
|
Hu Y, Qin S, Deng R. Impact of glioma metabolism-related gene ALPK1 on tumor immune heterogeneity and the regulation of the TGF-β pathway. Front Immunol 2025; 15:1512491. [PMID: 39845963 PMCID: PMC11753219 DOI: 10.3389/fimmu.2024.1512491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background Recent years have seen persistently poor prognoses for glioma patients. Therefore, exploring the molecular subtyping of gliomas, identifying novel prognostic biomarkers, and understanding the characteristics of their immune microenvironments are crucial for improving treatment strategies and patient outcomes. Methods We integrated glioma datasets from multiple sources, employing Non-negative Matrix Factorization (NMF) to cluster samples and filter for differentially expressed metabolic genes. Additionally, we utilized Weighted Gene Co-expression Network Analysis (WGCNA) to identify key genes. A predictive model was developed utilizing the optimal consistency index derived from a combination of 101 machine learning techniques, and its effectiveness was confirmed through multiple datasets employing different methodologies. In-depth analyses were conducted on immune cell infiltration and tumor microenvironmental aspects. Single-cell sequencing data were employed for clustering and differential expression analysis of genes associated with glioma. Finally, the immune relevance of the model gene ALPK1 in the context of pan-cancer was explored, including its relationship with immune checkpoints. Results The application of NMF, coupled with differential analysis of metabolic-related genes, led to the identification of two clusters exhibiting significant differences in survival, age, and metabolic gene expression among patients. Core genes were identified through WGCNA, and a total of 101 machine learning models were constructed, with LASSO+GBM selected as the optimal model, demonstrating robust validation performance. Comprehensive analyses revealed that high-risk groups exhibited greater expression of specific genes, with ALPK1 showing significant correlations with immune regulation. Conclusion This research employed a multi-dataset strategy and various methods to clarify the differences in metabolic traits and immune conditions in glioma patients, while creating an innovative prognostic risk evaluation framework. These results offer fresh perspectives on the intricate biological processes that define gliomas.
Collapse
Affiliation(s)
- YaoFeng Hu
- Department of Neurological Care Unit, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - Sen Qin
- Department of Orthopedics, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - RuCui Deng
- Department of Neurological Care Unit, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| |
Collapse
|
4
|
Xiang Z, Wang Y, Ma X, Song S, He Y, Zhou J, Feng L, Yang S, Wu Y, Yu B, Xia G, Xu W, Zhao Y, Wang L. Targeting the NOTCH2/ADAM10/TCF7L2 Axis-Mediated Transcriptional Regulation of Wnt Pathway Suppresses Tumor Growth and Enhances Chemosensitivity in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405758. [PMID: 39601111 PMCID: PMC11744699 DOI: 10.1002/advs.202405758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/12/2024] [Indexed: 11/29/2024]
Abstract
Wnt/β-catenin/transcription factor (TCF) transcriptional activity plays an integral role in colorectal cancer (CRC) carcinogenesis. However, to date, no drugs targeting this pathway are used in clinical practice owing to the undesirable and serious side effects. In this study, it is found that the transcriptional regulation of Wnt pathway is activated and associated with liver metastasis in CRC. Through high-throughput screening of 24 inhibitors on 12 CRC and three colorectal organoids in this organoid living biobank, adavivint is found to exhibit anti-tumor activity and low toxicity in colorectal organoids, independent of the canonical Wnt/β-catenin signaling. Mechanistically, ADAM10 is screened as a target of adavivint to specifically regulate the protein expression of NOTCH2, which mediates the transcriptional regulation of the Wnt pathway. NOTCH2 not directly interact with TCF7-like 2 (TCF7L2), a key downstream transcriptional factor of canonical Wnt/β-catenin signaling, but directly activated the transcription of TCF7L2 and Wnt target genes, such as MYC, JUN and CCND1/2. Furthermore, use of adavivint or blockage of ADAM10/NOTCH2/TCF7L2 signaling enhances the chemosensitivity of CRC cells. Overall, this study provides a promising candidate for the development of small-molecule inhibitors and reveals a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhen Xiang
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Yiwei Wang
- Department of general surgeryShanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RdShanghai200233P. R. China
| | - Xiao Ma
- Fudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032P. R. China
| | - Shuzheng Song
- Department of Colorectal SurgeryDepartment of General SurgeryShanghai East HospitalTongji University School of Medicine150 Jimo RoadShanghai200120P. R. China
| | - Yuanqiao He
- Center of Laboratory Animal ScienceNanchang UniversityNo.999, Xuefu RoadNanchang330031P. R. China
| | - Jiamin Zhou
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Longhai Feng
- Department of Colorectal SurgeryThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)1 Banshan East RoadHangzhou310022P. R. China
| | - Su Yang
- Department of Thoracic SurgeryRuijin HospitalShanghai Jiaotong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yibin Wu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Bingran Yu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Guangkai Xia
- Department of general surgeryShanghai Jiao Tong University Affiliated Sixth People's Hospital600 Yishan RdShanghai200233P. R. China
| | - Weiqi Xu
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Yiming Zhao
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| | - Lu Wang
- Department of Hepatic SurgeryFudan University Shanghai Cancer Center270 Dong‐An RoadShanghai200032China
| |
Collapse
|
5
|
Yang R, Qi Y, Zhang X, Gao H, Yu Y. Living biobank: Standardization of organoid construction and challenges. Chin Med J (Engl) 2024; 137:3050-3060. [PMID: 39663560 PMCID: PMC11706585 DOI: 10.1097/cm9.0000000000003414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Indexed: 12/13/2024] Open
Abstract
ABSTRACT In multiple areas such as science, technology, and economic activities, it is necessary to unify the management of repetitive tasks or concepts by standardization to obtain the best order and high efficiency. Organoids, as living tissue models, have rapidly developed in the past decade. Organoids can be used repetitively for in vitro culture, cryopreservation, and recovery for further utilization. Because organoids can recapitulate the parental tissues' morphological phenotypes, cell functions, biological behaviors, and genomic profiles, they are known as renewable "living biobanks". Organoids cover two mainstream fields: Adult stem cell-derived organoids (also known as patient-derived organoids) and induced pluripotent stem cell-derived and/or embryonic stem cell-derived organoids. Given the increasing importance of organoids in the development of new drugs, standardized operation, and management in all steps of organoid construction is an important guarantee to ensure the high quality of products. In this review, we systematically introduce the standardization of organoid construction operation procedures, the standardization of laboratory construction, and available standardization documents related to organoid culture that have been published so far. We also proposed the challenges and prospects in this field.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yao Qi
- National Engineering Center for Biochip at Shanghai, Shanghai 200120, China
| | - Xiaoyan Zhang
- National Engineering Center for Biochip at Shanghai, Shanghai 200120, China
| | - Hengjun Gao
- National Engineering Center for Biochip at Shanghai, Shanghai 200120, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
6
|
Co IL, Fomina A, Nurse M, McGuigan AP. Applications and evolution of 3D cancer-immune cell models. Trends Biotechnol 2024; 42:1615-1627. [PMID: 39025680 DOI: 10.1016/j.tibtech.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024]
Abstract
Understanding the highly complex tumor-immune landscape is an important goal for developing novel immune therapies for solid cancers. To this end, 3D cancer-immune models have emerged as patient-relevant in vitro tools for modeling the tumor-immune landscape and the cellular interactions within it. In this review, we provide an overview of the components and applications of 3D cancer-immune models and discuss their evolution from 2015 to 2023. Specifically, we observe trends in primary cell-sourced, T cell-based complex models used for therapy evaluation and biological discovery. Finally, we describe the challenges of implementing 3D cancer-immune models and the opportunities for maximizing their potential for deciphering the complex tumor-immune microenvironment and identifying novel, clinically relevant drug targets.
Collapse
Affiliation(s)
- Ileana L Co
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada
| | - Aleksandra Fomina
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada
| | - Michelle Nurse
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St., Toronto, ON, M5S 3E5, Canada
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St., Toronto, ON, M5S 3E5, Canada.
| |
Collapse
|
7
|
Xu J, Xu X, Zhang H, Wu J, Pan R, Zhang B. Tumor-associated inflammation: The role and research progress in tumor therapy. J Drug Deliv Sci Technol 2024; 102:106376. [DOI: 10.1016/j.jddst.2024.106376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Taş İ, Varlı M, Pulat S, Sim HB, Kim JJ, Kim H. TDO2 inhibition counters Benzo[a]pyrene-induced immune evasion and suppresses tumorigenesis in lung adenocarcinoma. Cancer Metab 2024; 12:36. [PMID: 39593177 PMCID: PMC11590479 DOI: 10.1186/s40170-024-00365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
INTRODUCTION Benzo[a]pyrene (BaP) is a toxic polycyclic aromatic hydrocarbon known as an exogenous AhR ligand. This study investigates the role of BaP in inducing immune checkpoint expression in lung adenocarcinoma (LUAD) and the underlying mechanisms involving the aryl hydrocarbon receptor (AhR) and tryptophan (Trp) metabolism. METHODS We assessed the expression of immune checkpoint molecules, including PD-L1 and ICOSL, in lung epithelial cell lines (BEAS-2B and H1975) exposed to BaP. The involvement of AhR in BaP-induced immune checkpoint expression was examined using AhR silencing (siAhR). Additionally, the role of Trp metabolism in BaP-mediated immune evasion was explored through culturing in Trp (-/+) condition media, treatments with the inhibitors of rate-limiting enzymes in Trp metabolism (TDO2 and IDO1) and analyses of Trp-catabolizing enzymes. The therapeutic potential of targeting Trp metabolism, specifically TDO2, was evaluated in vivo using C57BL/6 mice orthotopically inoculated with LUAD cells. RESULTS BaP exposure significantly upregulated the mRNA and surface expression of PD-L1 and ICOSL, with AhR playing a crucial role in this induction. Trp metabolism was found to enhance BaP-mediated immune evasion, as indicated by stronger induction of immune checkpoints in Trp (+) media and the upregulation of Trp-catabolizing enzymes. TDO2 inhibition markedly suppressed the surface expression of PD-L1 and ICOSL, demonstrating the importance of Trp metabolism in BaP-induced immune evasion. Further analysis confirmed the high TDO2 expression in lung adenocarcinoma and its association with poor patient survival. Using an orthotopic implantation mouse model, we demonstrated the inhibitory effect of two different TDO2 inhibitors on tumorigenesis, immune checkpoints, and tryptophan metabolism. CONCLUSIONS This study highlights the key mechanisms behind BaP-induced immune evasion in LUAD, particularly through the TDO2/AhR axis. It reveals how TDO2 inhibitors can counteract immune checkpoint activation and boost anti-tumor immunity, suggesting new paths for targeted lung cancer immunotherapy. The findings significantly improve our understanding of immune evasion in LUAD and underscore the therapeutic promise of TDO2 inhibition.
Collapse
Affiliation(s)
- İsa Taş
- College of Pharmacy, Sunchon National University, Sunchon, 57922, Jeonnam, Republic of Korea.
| | - Mücahit Varlı
- College of Pharmacy, Sunchon National University, Sunchon, 57922, Jeonnam, Republic of Korea
| | - Sultan Pulat
- College of Pharmacy, Sunchon National University, Sunchon, 57922, Jeonnam, Republic of Korea
| | - Hyun Bo Sim
- Department of Biomedical Science, Sunchon National University, Sunchon, 57922, Jeonnam, Republic of Korea
| | - Jong-Jin Kim
- Department of Biomedical Science, Sunchon National University, Sunchon, 57922, Jeonnam, Republic of Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon, 57922, Jeonnam, Republic of Korea.
| |
Collapse
|
9
|
Li Y, Liao W, Sun L. Application of tumor organoids simulating the tumor microenvironment in basic and clinical research of tumor immunotherapy. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:1316-1326. [PMID: 39788520 PMCID: PMC11628225 DOI: 10.11817/j.issn.1672-7347.2024.240187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Indexed: 01/12/2025]
Abstract
Immunotherapy has led to groundbreaking advances in anti-tumor treatment, yet significant clinical challenges remain such as the low proportion of beneficiaries and the lack of effective platforms for predicting therapeutic response. Organoid technology provides a novel solution to these issues. Organoids are three-dimensional tissue cultures derived from adult stem cells or pluripotent stem cells that closely replicate the structural and biological characteristics of native organs, demonstrating particularly strong potential in modeling the tumor microenvironment (TME). Tumor organoids can simulate TME effectively by retaining endogenous matrix components, including various immune cells, or by adding immune cells, cancer-associated fibroblasts, and other components. This provides a novel platform for predicting immunotherapy outcomes, evaluating adoptive cell therapies, and selecting personalized treatment options for patients. Summarizing strategies for constructing tumor organoids that simulate the microenvironment and understanding their advancements in immunotherapy research and clinical application can provide new insights for the development of tumor immunotherapy.
Collapse
Affiliation(s)
- Yizheng Li
- Cancer Center, Xiangya Hospital, Central South University, Hunan Key Laboratory of Molecular Radiation Oncology, International Cooperation Base in Science and Technology for Cancer Precision Medicine, National Clinical Research Center for Geriatric Disorders, Changsha 410008.
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lunquan Sun
- Cancer Center, Xiangya Hospital, Central South University, Hunan Key Laboratory of Molecular Radiation Oncology, International Cooperation Base in Science and Technology for Cancer Precision Medicine, National Clinical Research Center for Geriatric Disorders, Changsha 410008.
| |
Collapse
|
10
|
Yang R, Kwan W, Du Y, Yan R, Zang L, Li C, Zhu Z, Cheong IH, Kozlakidis Z, Yu Y. Drug-induced senescence by aurora kinase inhibitors attenuates innate immune response of macrophages on gastric cancer organoids. Cancer Lett 2024; 598:217106. [PMID: 38992487 PMCID: PMC11364160 DOI: 10.1016/j.canlet.2024.217106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
Diffuse-type gastric cancer (DGC) is a subtype of gastric cancer with aggressiveness and poor prognosis. It is of great significance to find sensitive drugs for DGC. In the current study, a total of 20 patient-derived organoids (PDOs) were analyzed for screening the therapeutic efficacy of small molecule kinases inhibitors on gastric cancers, especially the therapeutic difference between intestinal-type gastric cancer (IGCs) and DGCs. The IGCs are sensitive to multiple kinases inhibitors, while DGCs are resistant to most of these kinases inhibitors. It was found that DGCs showed drug-induced senescent phenotype after treatment by aurora kinases inhibitors (AURKi) Barasertib-HQPA and Danusertib. The cell diameter of cancer cells are increased with stronger staining of senescence-associated β-galactosidase (SA-β-GAL), and characteristic appearance of multinucleated giant cells. The senescent cancer cells secrete large amounts of chemokine MCP-1/CCL2, which recruit and induce macrophage to M2-type polarization in PDOs of DGC (DPDOs)-macrophage co-culture system. The up-regulation of local MCP-1/CCL2 can interact with MCP-1/CCL2 receptor (CCR2) expressed on macrophages and suppress their innate immunity to cancer cells. Overall, the special response of DGC to AURKi suggests that clinicians should select a sequential therapy with senescent cell clearance after AURKi treatment for DGC.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wingyan Kwan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yutong Du
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Ranlin Yan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Lu Zang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Chen Li
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Zhenggang Zhu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Io Hong Cheong
- Healthy Macau New-Generation Association, 999078, Macau, China
| | - Zisis Kozlakidis
- Laboratory Services and Biobank Group of International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366, LYON, CEDEX 07, France.
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
11
|
Yang R, Qi Y, Kwan W, Du Y, Yan R, Zang L, Yao X, Li C, Zhu Z, Zhang X, Gao H, Cheong IH, Kozlakidis Z, Yu Y. Paired organoids from primary gastric cancer and lymphatic metastasis are useful for personalized medicine. J Transl Med 2024; 22:754. [PMID: 39135062 PMCID: PMC11318189 DOI: 10.1186/s12967-024-05512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Organoids are approved by the US FDA as an alternative to animal experiments to guide drug development and for sensitivity screening. Stable organoids models of gastric cancer are desirable for personalized medicine and drug screening. METHODS Tumor tissues from a primary cancer of the stomach and metastatic cancer of the lymph node were collected for 3D culture. By long-term culture for over 50 generations in vitro, we obtained stably growing organoid lines. We analyzed short tandem repeats (STRs) and karyotypes of cancer cells, and tumorigenesis of the organoids in nude mice, as well as multi-omics profiles of the organoids. A CCK8 method was used to determine the drugs sensitivity to fluorouracil (5-Fu), platinum and paclitaxel. RESULTS Paired organoid lines from primary cancer (SPDO1P) and metastatic lymph node (SPDO1LM) were established with unique STRs and karyotypes. The organoid lines resulted in tumorigenesis in vivo and had clear genetic profiles. Compared to SPDO1P from primary cancer, upregulated genes of SPDO1LM from the metastatic lymph node were enriched in pathways of epithelial-mesenchymal transition and angiogenesis with stronger abilities of cell migration, invasion, and pro-angiogenesis. Based on drug sensitivity analysis, the SOX regimen (5-Fu plus oxaliplatin) was used for chemotherapy with an optimal clinical outcome. CONCLUSIONS The organoid lines recapitulate the drug sensitivity of the parental tissues. The paired organoid lines present a step-change toward living biobanks for further translational usage.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yao Qi
- Shanghai Engineering Center for Molecular Medicine, Zhangjiang, Shanghai, 200120, China
| | - Wingyan Kwan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yutong Du
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ranlin Yan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lu Zang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuexin Yao
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chen Li
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhenggang Zhu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyan Zhang
- Shanghai Engineering Center for Molecular Medicine, Zhangjiang, Shanghai, 200120, China
| | - Hengjun Gao
- Shanghai Engineering Center for Molecular Medicine, Zhangjiang, Shanghai, 200120, China
| | - Io Hong Cheong
- Healthy Macau New-Generation Association, Macau, 999078, China
| | - Zisis Kozlakidis
- Laboratory Services and Biobank Group of International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, LYON CEDEX 07, CS 90627, 69366, France.
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
12
|
Tsai HC, Chen YH, Jen JY, Chang HM. Increased expression of the kynurenine pathway in mice with eosinophilic meningitis caused by Angiostrongylus cantonensis infection. Acta Trop 2024; 256:107251. [PMID: 38763319 DOI: 10.1016/j.actatropica.2024.107251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/12/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024]
Abstract
Angiostrongylus cantonensis is the major cause of eosinophilic meningitis worldwide. The imbalance of neurotoxic and neuroprotective metabolites in the kynurenine pathway (KP) have been suggested to contribute to the pathogenesis of central nervous system (CNS) infection. We hypothesized that KP may also be involved in parasitic eosinophilic meningitis. BALB/c mice were orally infected with 40 A. cantonensis L3, intraperitoneal dexamethasone at a dose of 500 µg/kg/day was administered from the seventh day of infection until the end of the study. The Evans blue method was used to analyze blood-brain barrier (BBB) dysfunction, and indoleamine 2,3-dioxygenase (IDO) proteins levels was measured by Western blot, immunohistochemistry (IHC), and immunofluorescence. Tryptophan and kynurenine concentrations were analyzed by IHC and liquid chromatography-tandem mass spectrometry (LC-MS/MS). The concentrations of Evans blue, IDO, tryptophan and kynurenine in the different groups of mice were compared using the nonparametric Kruskal-Wallis test. BBB dysfunction was found in mice with eosinophilic meningitis. The administration of dexamethasone significantly decreased the amount of Evans blue. An increased IDO expression was shown in Western blot, IHC and immunofluorescence following 2-3 weeks infection. Increased tryptophan and kynurenine expressions in the brain and cerebrospinal fluid (CSF) were also found in IHC and LC-MS/MS studies. The administration of dexamethasone significantly decreased the amount of IDO, tryptophan and kynurenine. In conclusion, A. cantonensis infection inducing BBB damage, then increased the influx of tryptophan into CSF. The administration of dexamethasone significantly decreased the amount of IDO, tryptophan and kynurenine.
Collapse
Affiliation(s)
- Hung-Chin Tsai
- Section of Infectious Diseases, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan and National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| | - Yu-Hsin Chen
- Section of Infectious Diseases, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan and National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jing-Yueh Jen
- College of Pharmacy and Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Hui-Min Chang
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
13
|
Liao Y, Xie X, Zhang C, Zhong H, Shan L, Yu P, Xu L. Quercetin exerts anti-tumor immune mechanism by regulating IL-6/JAK2/STAT3 signaling pathway to deplete Treg cells. Toxicon 2024; 243:107747. [PMID: 38714236 DOI: 10.1016/j.toxicon.2024.107747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/09/2024]
Abstract
Breast cancer is still the leading cause of death among women worldwide. Due to the lack of effective drug targets, triple-negative breast cancer has a worse prognosis and higher mortality compared with other types of breast cancer, and chemotherapy is still the main treatment for triple-negative breast cancer at present. Quercetin (QUE) is a flavonoid compound found in a variety of fruits and vegetables. The mechanism of QUE has been extensively studied, such as prostate cancer, colon cancer, ovarian cancer, etc. However, the anti-tumor immune mechanism of QUE in triple-negative breast cancer remains unclear. Therefore, we assessed the anti-tumor immune effects of QUE on triple-negative breast cancer using both 4T1 cells and a xenograft mouse model of 4T1 cells. In vitro, we examined the inhibitory effects of QUE on 4T1 cells and its molecular mechanisms through MTT, Transwell, ELISA, and Western blotting. In vivo, by establishing a xenograft mouse model, we utilized flow cytometry, immunohistochemistry, ELISA, and Western blotting to evaluate the anti-tumor immune effects of QUE on triple-negative breast cancer. The results indicate that QUE inhibits the proliferation, migration, and invasion of 4T1 cells, concurrently significantly suppressing the IL-6/JAK2/STAT3 signaling pathway. Furthermore, it depletes Treg cell content in 4T1 xenograft mice, thereby improving the tumor immune microenvironment and promoting the cytotoxicity of relevant tumor immune cells. These findings suggest that QUE may serve as a potential adjuvant for immune therapy in triple-negative breast cancer.
Collapse
Affiliation(s)
- Yupei Liao
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Xiaoqing Xie
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Chu Zhang
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Haijing Zhong
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Pei Yu
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Lipeng Xu
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
14
|
Liu J, Wang T, Zhang W, Huang Y, Wang X, Li Q. Association between Metabolic Reprogramming and Immune Regulation in Digestive Tract Tumors. Oncol Res Treat 2024; 47:273-286. [PMID: 38636467 DOI: 10.1159/000538659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/28/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND The cancers of the digestive tract, including colorectal cancer (CRC), gastric cancer, and esophageal cancer, are part of the most common cancers as well as one of the most important leading causes of cancer death worldwide. SUMMARY Despite the emergence of immune checkpoint inhibitors (e.g., anti-CTLA-4 and anti-PD-1/PD-L1) in the past decade, offering renewed optimism in cancer treatment, only a fraction of patients derive benefit from these therapies. This limited efficacy may stem from tumor heterogeneity and the impact of metabolic reprogramming on both tumor cells and immune cells within the tumor microenvironment (TME). The metabolic reprogramming of glucose, lipids, amino acids, and other nutrients represents a pivotal hallmark of cancer, serving to generate energy, reducing equivalent and biological macromolecule, thereby fostering tumor proliferation and invasion. Significantly, the metabolic reprogramming of tumor cells can orchestrate changes within the TME, rendering patients unresponsive to immunotherapy. KEY MESSAGES In this review, we predominantly encapsulate recent strides on metabolic reprogramming among digestive tract cancer, especially CRC, in the TME with a focus on how these alterations influence anti-tumor immunity. Additionally, we deliberate on potential strategies to address these abnormities in metabolic pathways and the viability of combined therapy within the realm of anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Jiafeng Liu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxin Huang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinhai Wang
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Abstract
Although there is little direct evidence supporting that stress affects cancer incidence, it does influence the evolution, dissemination and therapeutic outcomes of neoplasia, as shown in human epidemiological analyses and mouse models. The experience of and response to physiological and psychological stressors can trigger neurological and endocrine alterations, which subsequently influence malignant (stem) cells, stromal cells and immune cells in the tumour microenvironment, as well as systemic factors in the tumour macroenvironment. Importantly, stress-induced neuroendocrine changes that can regulate immune responses have been gradually uncovered. Numerous stress-associated immunomodulatory molecules (SAIMs) can reshape natural or therapy-induced antitumour responses by engaging their corresponding receptors on immune cells. Moreover, stress can cause systemic or local metabolic reprogramming and change the composition of the gastrointestinal microbiota which can indirectly modulate antitumour immunity. Here, we explore the complex circuitries that link stress to perturbations in the cancer-immune dialogue and their implications for therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Yuting Ma
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.
| | - Guido Kroemer
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Gu Z, Wu Q, Shang B, Zhang K, Zhang W. Organoid co-culture models of the tumor microenvironment promote precision medicine. CANCER INNOVATION 2024; 3:e101. [PMID: 38948532 PMCID: PMC11212345 DOI: 10.1002/cai2.101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 07/02/2024]
Abstract
In recent years, the three-dimensional (3D) culture system has emerged as a promising preclinical model for tumor research owing to its ability to replicate the tissue structure and molecular characteristics of solid tumors in vivo. This system offers several advantages, including high throughput, efficiency, and retention of tumor heterogeneity. Traditional Matrigel-submerged organoid cultures primarily support the long-term proliferation of epithelial cells. One solution for the exploration of the tumor microenvironment is a reconstitution approach involving the introduction of exogenous cell types, either in dual, triple or even multiple combinations. Another solution is a holistic approach including patient-derived tumor fragments, air-liquid interface, suspension 3D culture, and microfluidic tumor-on-chip models. Organoid co-culture models have also gained popularity for studying the tumor microenvironment, evaluating tumor immunotherapy, identifying predictive biomarkers, screening for effective drugs, and modeling infections. By leveraging these 3D culture systems, it is hoped to advance the clinical application of therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Zhaoru Gu
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Quanyou Wu
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Bingqing Shang
- Department of Urology, National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wen Zhang
- Department of Immunology, National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
17
|
Huang P, Sun R, Xu C, Jiang Z, Zuo M, Li Y, Liu R, Gong P, Han Y, Fang J, Li P, Shao C, Shi Y. Glucocorticoid activates STAT3 and NF-κB synergistically with inflammatory cytokines to enhance the anti-inflammatory factor TSG6 expression in mesenchymal stem/stromal cells. Cell Death Dis 2024; 15:70. [PMID: 38238297 PMCID: PMC10796730 DOI: 10.1038/s41419-024-06430-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/22/2024]
Abstract
Glucocorticoid (GC) is essential for maintaining immune homeostasis. While GC is known to regulate the expression of genes related to inflammation in immune cells, the effects of GC, especially in the presence of inflammation, on non-immune cells remain largely unexplored. In particular, the impact of GC on inflammatory cytokine-induced immune modulatory responses of tissue stromal cells is unknown, though it has been widely used to modulate tissue injuries. Here we found that GC could enhance the expression of TSG6, a vital tissue repair effector molecule, in IFNγ and TNFα treated human umbilical cord (UC)-MSCs. NF-κB activation was found to be required for GC-augmented TSG6 upregulation. STAT3, but not STAT1, was also found to be required for the TSG6 upregulation in MSCs exposed to IFNγ, TNFα and GC. Moreover, the phosphorylation (activation) of STAT3 was attenuated when NF-κB was knocked down. Importantly, human UC-MSCs pretreated with a cocktail containing GC, IFNγ, and TNFα could significantly enhance the therapeutic effect of human UC-MSCs in an acute lung injury mouse model, as reflected by reduced infiltration of immune cells and down-regulation of iNOS in macrophages in the lung. Together, the findings reveal a novel link between GR, NF-κB and STAT3 in regulating the immunomodulatory and regenerative properties of MSCs, providing novel information for the understanding and treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Peiqing Huang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Rongrong Sun
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Chenchang Xu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Zixuan Jiang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Muqiu Zuo
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Yinghong Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Pixia Gong
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Yuyi Han
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Peishan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China.
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Suzhou Medical College, Suzhou, China.
| |
Collapse
|
18
|
Yang R, Xiang Z, Yan R, Kwan W, Zang L, Zhu Z, Qi Y, Xu Y, Zhang X, Gao H, Yu Y. Benchmark for establishment of organoids from gastrointestinal epithelium and cancer based on available consumables and reagents. Chin J Cancer Res 2023; 35:636-644. [PMID: 38204440 PMCID: PMC10774133 DOI: 10.21147/j.issn.1000-9604.2023.06.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Gastrointestinal cancers are a public health problem that threatens the lives of human being. A good experimental model is a powerful tool to promote the uncovering pathogenesis and establish novel treatment methods. High-quality biomedical research requires experimental models to recapitulate the physiological and pathological states of their parental tissues as much as possible. Organoids are such experimental models. Organoids refer to small organ-like cellular clusters formed by the expansion and passaging of living tissues in 3D culture medium in vitro. Organoids are highly similar to the original tissues in terms of cellular composition, cell functions, and genomic profiling. Organoids have many advantages, such as short preparation cycles, long-term storage based on cryopreservation, and reusability. In recent years, researchers carried out the establishment of organoids from gastrointestinal mucosa and cancer tissues, and accumulated valuable experiences. In order to promote effective usage and further development of organoid-related technologies in the research of gastrointestinal diseases, this study proposes a benchmark based on utilization of available experimental consumables and reagents, which are involved in the key steps such as collection and pretreatment of biospecimen, organoid construction, organoid cryopreservation and recovery, growth status evaluation, and organoid quality control. We believe that the standard for the construction and preservation of organoids derived from human gastrointestinal epithelium and cancer tissues can provide an important reference for the majority of scientific researchers.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhen Xiang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ranlin Yan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wingyan Kwan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lu Zang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhenggang Zhu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yao Qi
- Shanghai Engineering Center for Molecular Medicine, Shanghai 200120, China
| | - Yanping Xu
- Shanghai Engineering Center for Molecular Medicine, Shanghai 200120, China
| | - Xiaoyan Zhang
- Shanghai Engineering Center for Molecular Medicine, Shanghai 200120, China
| | - Hengjun Gao
- Shanghai Engineering Center for Molecular Medicine, Shanghai 200120, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
19
|
Qin L, Wu J. Targeting anticancer immunity in oral cancer: Drugs, products, and nanoparticles. ENVIRONMENTAL RESEARCH 2023; 239:116751. [PMID: 37507044 DOI: 10.1016/j.envres.2023.116751] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 07/30/2023]
Abstract
Oral cavity carcinomas are the most frequent malignancies among head and neck malignancies. Oral tumors include not only oral cancer cells with different potency and stemness but also consist of diverse cells, containing anticancer immune cells, stromal and also immunosuppressive cells that influence the immune system reactions. The infiltrated T and natural killer (NK) cells are the substantial tumor-suppressive immune compartments in the tumor. The infiltration of these cells has substantial impacts on the response of tumors to immunotherapy, chemotherapy, and radiotherapy. Nevertheless, cancer cells, stromal cells, and some other compartments like regulatory T cells (Tregs), macrophages, and myeloid-derived suppressor cells (MDSCs) can repress the immune responses against malignant cells. Boosting anticancer immunity by inducing the immune system or repressing the tumor-promoting cells is one of the intriguing approaches for the eradication of malignant cells such as oral cancers. This review aims to concentrate on the secretions and interactions in the oral tumor immune microenvironment. We review targeting tumor stroma, immune system and immunosuppressive interactions in oral tumors. This review will also focus on therapeutic targets and therapeutic agents such as nanoparticles and products with anti-tumor potency that can boost anticancer immunity in oral tumors. We also explain possible future perspectives including delivery of various cells, natural products and drugs by nanoparticles for boosting anticancer immunity in oral tumors.
Collapse
Affiliation(s)
- Liling Qin
- Gezhouba Central Hospital of the Third Clinical Medical College of Three Gorges University, Yichang, Hubei, 443002, China
| | - Jianan Wu
- Experimental and Practical Teaching Center, Hubei College of Chinese Medicine, Jingzhou, Hubei, 434000, China.
| |
Collapse
|
20
|
Xie Z, Chen C, Ma’ayan A. Dex-Benchmark: datasets and code to evaluate algorithms for transcriptomics data analysis. PeerJ 2023; 11:e16351. [PMID: 37953774 PMCID: PMC10638921 DOI: 10.7717/peerj.16351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023] Open
Abstract
Many tools and algorithms are available for analyzing transcriptomics data. These include algorithms for performing sequence alignment, data normalization and imputation, clustering, identifying differentially expressed genes, and performing gene set enrichment analysis. To make the best choice about which tools to use, objective benchmarks can be developed to compare the quality of different algorithms to extract biological knowledge maximally and accurately from these data. The Dexamethasone Benchmark (Dex-Benchmark) resource aims to fill this need by providing the community with datasets and code templates for benchmarking different gene expression analysis tools and algorithms. The resource provides access to a collection of curated RNA-seq, L1000, and ChIP-seq data from dexamethasone treatment as well as genetic perturbations of its known targets. In addition, the website provides Jupyter Notebooks that use these pre-processed curated datasets to demonstrate how to benchmark the different steps in gene expression analysis. By comparing two independent data sources and data types with some expected concordance, we can assess which tools and algorithms best recover such associations. To demonstrate the usefulness of the resource for discovering novel drug targets, we applied it to optimize data processing strategies for the chemical perturbations and CRISPR single gene knockouts from the L1000 transcriptomics data from the Library of Integrated Network Cellular Signatures (LINCS) program, with a focus on understudied proteins from the Illuminating the Druggable Genome (IDG) program. Overall, the Dex-Benchmark resource can be utilized to assess the quality of transcriptomics and other related bioinformatics data analysis workflows. The resource is available from: https://maayanlab.github.io/dex-benchmark.
Collapse
Affiliation(s)
- Zhuorui Xie
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clara Chen
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Avi Ma’ayan
- Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Wang X, Ye X, Chen Y, Lin J. Mechanism of M2 type macrophage-derived extracellular vesicles regulating PD-L1 expression via the MISP/IQGAP1 axis in hepatocellular carcinoma immunotherapy resistance. Int Immunopharmacol 2023; 124:110848. [PMID: 37633233 DOI: 10.1016/j.intimp.2023.110848] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a prevailing cancer affecting human health. M2 macrophages are essential in mediating immune responses in tumors. This study investigated the action of M2 macrophages in immune escape of HCC. METHODS Mitotic spindle positioning (MISP), IQ motif containing GTPase activating protein 1 (IQGAP1) and programmed cell death-1 (PD-L1) levels in primary HCC/tumor-adjacent tissues were determined by Western blot, followed by correlation analysis. M2 macrophage and CD3+CD8+T cell percentages were estimated by flow cytometry. Hep3B and HepG2 cells were treated with M2 macrophage conditioned medium (M2-CM) and M2 macrophage-derived extracellular vesicles (M2-EVs) and/or co-cultured with CD8+T cells, followed by assessment of cell viability and apoptosis. TNF-α and INF-γ levels were measured by ELISA. MISP and IQGAP1 overexpression plasmids were transfected into HCC cells to explore their role in immune escape. The interactions among MISP, IQGAP1, STAT3, and PD-L1 were analyzed by co-immunoprecipitation. The mechanism of M2-EVs in HCC immune escape was verified in nude mice. RESULTS MISP/IQGAP1/PD-L1 were upregulated in HCC tissues. MISP negatively-correlated with IQGAP1/PD-L1 and IQGAP1 positively-correlated with PD-L1. M2 macrophages were reduced but CD8+T cells were increased in HCC tissues with high MISP expression. M2-CM or M2-EVs inhibited the killing ability of CD8+T cells, increased HCC cell viability, impeded HCC cell apoptosis, induced CD8+T cell apoptosis, downregulated TNF-α and INF-γ, and upregulated PD-L1. M2-EVs facilitated HCC cell immune escape by potentiating IQGAP1 nuclear translocation and activating STAT3 phosphorylation through MISP downregulation. In vivo experiments further verified the action of M2-EVs through MISP. CONCLUSION M2-EVs promote HCC cell immune escape by upregulating PD-L1 through the MISP/IQGAP1/STAT3 axis.
Collapse
Affiliation(s)
- Xiaobo Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Xuxing Ye
- Department of Traditional Chinese Medicine, Jinhua Municipal Central Hospital, 351 Mingyue Street, Wucheng District, Jinhua, 321001, China
| | - Yanping Chen
- Department of Gastroenterology, Jinhua Municipal Central Hospital, 351 Mingyue Street, Wucheng District, Jinhua, 321001, China
| | - Junmei Lin
- Department of Traditional Chinese Medicine, Jinhua Municipal Central Hospital, 351 Mingyue Street, Wucheng District, Jinhua, 321001, China.
| |
Collapse
|
22
|
Van Buren I, Madison C, Kohn A, Berry E, Kulkarni RP, Thompson RF. Survival Among Veterans Receiving Steroids for Immune-Related Adverse Events After Immune Checkpoint Inhibitor Therapy. JAMA Netw Open 2023; 6:e2340695. [PMID: 37906189 PMCID: PMC10618850 DOI: 10.1001/jamanetworkopen.2023.40695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/19/2023] [Indexed: 11/02/2023] Open
Abstract
Importance Systemic steroids are commonly used to manage immune-related adverse events (irAEs), but it remains unclear whether they may undermine immune checkpoint inhibitor (ICI) therapy outcomes. Few studies have assessed the impact of steroid timing and its association with continuation or cessation of ICI therapy. Objective To characterize how systemic steroids and steroid timing for irAEs are associated with survival in patients receiving ICI therapy. Design, Setting, and Participants This multicenter retrospective cohort study encompassed veterans receiving ICI for cancer between January 1, 2010, and December 31, 2021. Data analysis was conducted September 8, 2023. Exposures Identifiable primary diagnosis of cancer. Patients were categorized into 3 cohorts: those receiving no steroids, systemic steroids for irAEs, and steroids for non-irAE-associated reasons. All eligible patients received 1 or more doses of an ICI (atezolizumab, avelumab, cemiplimab, durvalumab, ipilimumab, nivolumab, or pembrolizumab). Eligible patients in the steroid group received at least 1 dose (intravenous, intramuscular, or oral) of dexamethasone, hydrocortisone, methylprednisolone, prednisone, or prednisolone. Steroid use at baseline for palliation or infusion prophylaxis or delivered as a single dose was deemed to be non-irAE associated. All other patterns of steroid use were assumed to be for irAEs. Main Outcomes and Measures The primary outcome was overall survival, with a 5-year follow-up after ICI initiation. Kaplan-Meier survival analyses were performed with pairwise log-rank tests to determine significance. Risk was modeled with Cox proportional hazard regression. Results The cohort consisted of 20 163 veterans receiving ICI therapy including 12 221 patients (mean [SD] age, 69.5 [8.0] years; 11 830 male patients [96.8%]; 9394 White patients [76.9%]) who received systemic steroids during ICI treatment and 7942 patients (mean [SD] age, 70.3 [8.5] years; 7747 male patients [97.5%]; 6085 White patients [76.6%]) who did not. Patients with an irAE diagnosis had significantly improved overall survival (OS) compared with those without (median [IQR] OS, 17.4 [6.6 to 48.5] months vs 10.5 [3.5 to 36.8] months; adjusted hazard ratio, 0.84; 95% CI, 0.81-0.84; P < .001). For patients with irAEs, systemic steroids for irAEs were associated with significantly improved survival compared with those who received steroids for non-irAE-related reasons or no steroid treatment (median [IQR] OS, 21.3 [9.3 to 58.2] months vs 13.6 [5.5 to 33.7] months vs 15.8 [4.9 to not reached] months; P <.001). However, among those who received steroids for irAEs, early steroid use (<2 months after ICI initiation) was associated with reduced relative survival benefit vs later steroid use, regardless of ICI continuation or cessation following steroid initiation (median [IQR] OS after ICI cessation 4.4 [1.9 to 19.5] months vs 16.0 [8.0 to 42.2] months; median [IQR] OS after ICI continuation, 16.0 [7.1 to not reached] months vs 29.2 [16.5 to 53.5] months; P <.001). Conclusions and Relevance This study suggests that steroids for irAE management may not abrogate irAE-associated survival benefits. However, early steroid administration within 2 months of ICI initiation is associated with shorter survival despite continuation of ICI therapy.
Collapse
Affiliation(s)
- Inga Van Buren
- Graduate Medical Education, St Joseph’s Medical Center, Stockton, California
| | - Cecelia Madison
- Research and Development, VA Portland Healthcare System, Portland, Oregon
| | - Aimee Kohn
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland
| | - Elizabeth Berry
- Department of Dermatology, Oregon Health & Science University, Portland
| | - Rajan P. Kulkarni
- Department of Dermatology, Oregon Health & Science University, Portland
- Operative Care Division, VA Portland Healthcare System, Portland, Oregon
| | - Reid F. Thompson
- Department of Radiation Medicine, Oregon Health & Science University, Portland
- Division of Hospital and Specialty Medicine, VA Portland Healthcare System, Portland, Oregon
| |
Collapse
|
23
|
Mao D, Zhou Z, Chen H, Liu X, Li D, Chen X, He Y, Liu M, Zhang C. Pleckstrin-2 promotes tumour immune escape from NK cells by activating the MT1-MMP-MICA signalling axis in gastric cancer. Cancer Lett 2023; 572:216351. [PMID: 37591356 DOI: 10.1016/j.canlet.2023.216351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/19/2023]
Abstract
Immune escape is a major challenge in tumour immunotherapy. Pleckstrin-2(PLEK2) plays a critical role in tumour progression, but its role in immune escape in gastric cancer (GC) remains uncharacterized. RNA sequencing was used to explore the differentially expressed genes in a GC cell line that was resistant to the antitumor effect of Natural killer (NK) cells. Apoptosis and the expression of IFN-γ and TNF-α were detected by flow cytometry (FCM). PLEK2 expression was examined by Western blotting and immunohistochemistry (IHC). PLEK2 was upregulated in MGC803R cells that were resistant to the antitumor effect of NK cells. PLEK2 knockout increased the sensitivity of GC cells to NK cell killing. PLEK2 expression was negatively correlated with MICA and positively correlated with MT1-MMP expression both in vitro and in vivo. PLEK2 promoted Sp1 phosphorylation through the PI3K-AKT pathway, thereby upregulating MT1-MMP expression, which ultimately led to MICA shedding. In mouse xenograft models, PLEK2 knockout inhibited intraperitoneal metastasis of GC cells and promoted NK cell infiltration. In summary, PLEK2 suppressed NK cell immune surveillance by promoting MICA shedding, which serves as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Deli Mao
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Zhijun Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, United States
| | - Hengxing Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Xinran Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Dongsheng Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Xiancong Chen
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Department of Gastrointestinal Surgery of the First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
24
|
Wu Y, Du B, Lin M, Ji X, Lv C, Lai J. The identification of genes associated T-cell exhaustion and construction of prognostic signature to predict immunotherapy response in lung adenocarcinoma. Sci Rep 2023; 13:13415. [PMID: 37592010 PMCID: PMC10435542 DOI: 10.1038/s41598-023-40662-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/16/2023] [Indexed: 08/19/2023] Open
Abstract
T-cell exhaustion (Tex) is considered to be a reason for immunotherapy resistance and poor prognosis in lung adenocarcinoma. Therefore, we used weighted correlation network analysis to identify Tex-related genes in the cancer genome atlas (TCGA). Unsupervised clustering approach based on Tex-related genes divided patients into cluster 1 and cluster 2. Then, we utilized random forest and the least absolute shrinkage and selection operator to identify nine key genes to construct a riskscore. Patients were classified as low or high-risk groups. The multivariate cox analysis showed the riskscore was an independent prognostic factor in TCGA and GSE72094 cohorts. Moreover, patients in cluster 2 with high riskscore had the worst prognosis. The immune response prediction analysis showed the low-risk group had higher immune, stromal, estimate scores, higher immunophenscore (IPS), and lower tumor immune dysfunction and exclusion score which suggested a better response to immune checkpoint inhibitors (ICIs) therapy in the low-risk group. In the meantime, we included two independent immunotherapy cohorts that also confirmed a better response to ICIs treatment in the low-risk group. Besides, we discovered differences in chemotherapy and targeted drug sensitivity between two groups. Finally, a nomogram was built to facilitate clinical decision making.
Collapse
Affiliation(s)
- Yahua Wu
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Bin Du
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Mingqiang Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Jin'an District, Fuzhou, 350000, Fujian, China
| | - Xiaohui Ji
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Chengliu Lv
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China
| | - Jinhuo Lai
- Department of Medical Oncology, Fujian Medical University Union Hospital, No. 29 Xinquan Street, Fuzhou, 350000, Fujian, China.
| |
Collapse
|
25
|
Lin Q, Wang X, Hu Y. The opportunities and challenges in immunotherapy: Insights from the regulation of PD-L1 in cancer cells. Cancer Lett 2023:216318. [PMID: 37454966 DOI: 10.1016/j.canlet.2023.216318] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
The immunosuppressive molecule programmed death-ligand 1 (PD-L1) is frequently upregulated in human cancers. Binding of PD-L1 to its receptor, programmed death-1 (PD-1), on activated T cells facilitates cancer cells to evade the host immune system. Antibody-based PD-1/PD-L1 inhibitors can inhibit PD-1/PD-L1 interaction allowing reactivate cytotoxic T cells to eradicate advanced cancer cells. However, the majority of cancer patients fail to respond to anti-PD-1/PD-L1 therapies and the molecular mechanisms for this remain poorly understood. Recent studies show that PD-L1 expression level on tumor cells affect the clinical efficacy of immune checkpoint therapies. Thus, furthering our understanding of the regulatory mechanisms of PD-L1 expression in cancer cells will be critical to improve clinical response rates and the efficacy of PD-1/PD-L1 immune therapies. Here we review recent studies, primarily focusing on the mechanisms that regulate PD-L1 expression at the transcriptional, post-transcriptional and protein level, with the purpose to drive the development of more targeted and effective anti-PD-1/PD-L1 cancer therapies.
Collapse
Affiliation(s)
- Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China.
| |
Collapse
|
26
|
Kiriyama Y, Nochi H. Regulation of PD-L1 Expression by Nuclear Receptors. Int J Mol Sci 2023; 24:9891. [PMID: 37373038 DOI: 10.3390/ijms24129891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The suppression of excessive immune responses is necessary to prevent injury to the body, but it also allows cancer cells to escape immune responses and proliferate. Programmed cell death 1 (PD-1) is a co-inhibitory molecule that is present on T cells and is the receptor for programmed cell death ligand 1 (PD-L1). The binding of PD-1 to PD-L1 leads to the inhibition of the T cell receptor signaling cascade. PD-L1 has been found to be expressed in many types of cancers, such as lung, ovarian, and breast cancer, as well as glioblastoma. Furthermore, PD-L1 mRNA is widely expressed in normal peripheral tissues including the heart, skeletal muscle, placenta, lungs, thymus, spleen, kidney, and liver. The expression of PD-L1 is upregulated by proinflammatory cytokines and growth factors via a number of transcription factors. In addition, various nuclear receptors, such as androgen receptor, estrogen receptor, peroxisome-proliferator-activated receptor γ, and retinoic-acid-related orphan receptor γ, also regulate the expression of PD-L1. This review will focus on the current knowledge of the regulation of PD-L1 expression by nuclear receptors.
Collapse
Affiliation(s)
- Yoshimitsu Kiriyama
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 769-2193, Kagawa, Japan
- Institute of Neuroscience, Tokushima Bunri University, Tokushima 769-2193, Kagawa, Japan
| | - Hiromi Nochi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 769-2193, Kagawa, Japan
| |
Collapse
|
27
|
Stone TW, Williams RO. Interactions of IDO and the Kynurenine Pathway with Cell Transduction Systems and Metabolism at the Inflammation-Cancer Interface. Cancers (Basel) 2023; 15:cancers15112895. [PMID: 37296860 DOI: 10.3390/cancers15112895] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023] Open
Abstract
The mechanisms underlying a relationship between inflammation and cancer are unclear, but much emphasis has been placed on the role of tryptophan metabolism to kynurenine and downstream metabolites, as these make a substantial contribution to the regulation of immune tolerance and susceptibility to cancer. The proposed link is supported by the induction of tryptophan metabolism by indoleamine-2,3-dioxygenase (IDO) or tryptophan-2,3-dioxygenase (TDO), in response to injury, infection or stress. This review will summarize the kynurenine pathway and will then focus on the bi-directional interactions with other transduction pathways and cancer-related factors. The kynurenine pathway can interact with and modify activity in many other transduction systems, potentially generating an extended web of effects other than the direct effects of kynurenine and its metabolites. Conversely, the pharmacological targeting of those other systems could greatly enhance the efficacy of changes in the kynurenine pathway. Indeed, manipulating those interacting pathways could affect inflammatory status and tumor development indirectly via the kynurenine pathway, while pharmacological modulation of the kynurenine pathway could indirectly influence anti-cancer protection. While current efforts are progressing to account for the failure of selective IDO1 inhibitors to inhibit tumor growth and to devise means of circumventing the issue, it is clear that there are wider factors involving the relationship between kynurenines and cancer that merit detailed consideration as alternative drug targets.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Richard O Williams
- The Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| |
Collapse
|
28
|
Verheijden RJ, van Eijs MJM, May AM, van Wijk F, Suijkerbuijk KPM. Immunosuppression for immune-related adverse events during checkpoint inhibition: an intricate balance. NPJ Precis Oncol 2023; 7:41. [PMID: 37173424 PMCID: PMC10182067 DOI: 10.1038/s41698-023-00380-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed perspectives for patients with cancer, but come with severe immune-related adverse events (irAEs). To prevent fatality or chronicity, these irAEs are often promptly treated with high-dose immunosuppressants. Until recently, evidence on the effects of irAE management on ICI efficacy has been sparse. As a result, algorithms for irAE management are mostly expert-opinion based and barely consider possible detrimental effects of immunosuppressants on ICI efficacy. However, recent growing evidence suggests that vigorous immunosuppressive management of irAEs comes with unfavourable effects on ICI efficacy and survival. With expansion of the indications of ICIs, evidence-based treatment of irAEs without hampering tumour control becomes more and more important. In this review, we discuss novel evidence from pre-clinical and clinical studies on the effects of different irAE management regimens including corticosteroids, TNF inhibition and tocilizumab on cancer control and survival. We provide recommendations for pre-clinical research, cohort studies and clinical trials that can help clinicians in tailored irAE management, minimising patients' burden while maintaining ICI efficacy.
Collapse
Affiliation(s)
- Rik J Verheijden
- Department of Medical Oncology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Mick J M van Eijs
- Department of Medical Oncology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
29
|
Ding JT, Yang KP, Zhou HN, Huang YF, Li H, Zong Z. Landscapes and mechanisms of CD8 + T cell exhaustion in gastrointestinal cancer. Front Immunol 2023; 14:1149622. [PMID: 37180158 PMCID: PMC10166832 DOI: 10.3389/fimmu.2023.1149622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
CD8+ T cells, a cytotoxic T lymphocyte, are a key component of the tumor immune system, but they enter a hyporeactive T cell state in long-term chronic inflammation, and how to rescue this depleted state is a key direction of research. Current studies on CD8+ T cell exhaustion have found that the mechanisms responsible for their heterogeneity and differential kinetics may be closely related to transcription factors and epigenetic regulation, which may serve as biomarkers and potential immunotherapeutic targets to guide treatment. Although the importance of T cell exhaustion in tumor immunotherapy cannot be overstated, studies have pointed out that gastric cancer tissues have a better anti-tumor T cell composition compared to other cancer tissues, which may indicate that gastrointestinal cancers have more promising prospects for the development of precision-targeted immunotherapy. Therefore, the present study will focus on the mechanisms involved in the development of CD8+ T cell exhaustion, and then review the landscapes and mechanisms of T cell exhaustion in gastrointestinal cancer as well as clinical applications, which will provide a clear vision for the development of future immunotherapies.
Collapse
Affiliation(s)
- Jia-Tong Ding
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Kang-Ping Yang
- The Second Clinical Medicine School, Nanchang University, Nanchang, China
| | - Hao-Nan Zhou
- Queen Mary School, Nanchang University, Nanchang, China
| | - Ying-Feng Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
Yang R, Yu Y. Patient-derived organoids in translational oncology and drug screening. Cancer Lett 2023; 562:216180. [PMID: 37061121 DOI: 10.1016/j.canlet.2023.216180] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Patient-derived organoids (PDO) are a new biomedical research model that can reconstruct phenotypic and genetic characteristics of the original tissue and are useful for research on pathogenesis and drug screening. To introduce the progression in this field, we review the key factors of constructing organoids derived from epithelial tissues and cancers, covering culture medium and matrix, morphological characteristics, genetic profiles, high-throughput drug screening, and application potential. We also discuss the co-culture system of cancer organoids with tumor microenvironment (TME) associated cells. The co-culture system is widely used in evaluating crosstalk of cancer cells with TME components, such as fibroblasts, endothelial cells, immune cells, and microorganisms. The article provides a prospective for standardized cultivation mode, automatic morphological evaluation, and drug sensitivity screening using high-throughput methods.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
31
|
Mu P, Zhou S, Lv T, Xia F, Shen L, Wan J, Wang Y, Zhang H, Cai S, Peng J, Hua G, Zhang Z. Newly developed 3D in vitro models to study tumor-immune interaction. J Exp Clin Cancer Res 2023; 42:81. [PMID: 37016422 PMCID: PMC10074642 DOI: 10.1186/s13046-023-02653-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 04/06/2023] Open
Abstract
Immunotherapy as a rapidly developing therapeutic approach has revolutionized cancer treatment and revitalized the field of tumor immunology research. 3D in vitro models are emerging as powerful tools considering their feature to maintain tumor cells in a near-native state and have been widely applied in oncology research. The novel 3D culture methods including the co-culture of organoids and immune cells, ALI culture, 3D-microfluidic culture and 3D-bioprinting offer new approaches for tumor immunology study and can be applied in many fields such as personalized treatment, immunotherapy optimizing and adoptive cell therapy. In this review, we introduce commonly used 3D in vitro models and summarize their applications in different aspects of tumor immunology research. We also provide a preliminary analysis of the current shortcomings of these models and the outlook of future development.
Collapse
Affiliation(s)
- Peiyuan Mu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Shujuan Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Tao Lv
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China
| | - Sanjun Cai
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Cancer institute, Fudan University Shanghai Cancer Center, 200032, Shanghai, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, 200032, Shanghai, China
| | - Junjie Peng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Fudan University, 200032, Shanghai, China
| | - Guoqiang Hua
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
- Cancer institute, Fudan University Shanghai Cancer Center, 200032, Shanghai, China.
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, 200032, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, 200032, China.
| |
Collapse
|
32
|
Dinarello A, Mills TS, Tengesdal IW, Powers NE, Azam T, Dinarello CA. Dexamethasone and OLT1177 Cooperate in the Reduction of Melanoma Growth by Inhibiting STAT3 Functions. Cells 2023; 12:294. [PMID: 36672229 PMCID: PMC9856388 DOI: 10.3390/cells12020294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
The NLRP3 inflammasome is a multimolecular complex that processes inactive IL-1β and IL-18 into proinflammatory cytokines. OLT1177 is an orally active small compound that specifically inhibits NLRP3. Here, B16F10 melanoma were implanted in mice and treated with OLT1177 as well as combined with the glucocorticoid dexamethasone. At sacrifice, OLT1177 treated mice had significantly smaller tumors compared to tumor-bearing mice treated with vehicle. However, the combined treatment of OLT1177 plus dexamethasone revealed a greater suppression of tumor growth. This reduction was accompanied by a downregulation of nuclear and mitochondrial STAT3-dependent gene transcription and by a significant reduction of STAT3 Y705 and S727 phosphorylations in the tumors. In vitro, the human melanoma cell line 1205Lu, stimulated with IL-1α, exhibited significantly lower levels of STAT3 Y705 phosphorylation by the combination treatment, thus affecting the nuclear functions of STAT3. In the same cells, STAT3 serine 727 phosphorylation was also lower, affecting the mitochondrial functions of STAT3. In addition, metabolic analyses revealed a marked reduction of ATP production rate and glycolytic reserve in cells treated with the combination of OLT1177 plus dexamethasone. These findings demonstrate that the combination of OLT1177 and dexamethasone reduces tumor growth by targeting nuclear as well as mitochondrial functions of STAT3.
Collapse
Affiliation(s)
- Alberto Dinarello
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Taylor S. Mills
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Isak W. Tengesdal
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Nicholas E. Powers
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Tania Azam
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado, Aurora, Denver, CO 80045, USA
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
33
|
Cui Y, Han X, Liu H, Xie Q, Guan Y, Yin B, Xiao J, Feng D, Wang X, Li J, Chen J, Liu X, Li X, Nie W, Ma L, Liu H, Liang J, Li Y, Wang B, Wang J. Impact of endogenous glucocorticoid on response to immune checkpoint blockade in patients with advanced cancer. Front Immunol 2023; 14:1081790. [PMID: 37114049 PMCID: PMC10126286 DOI: 10.3389/fimmu.2023.1081790] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Background Previous studies indicate that exogenous use of glucocorticoid (GC) affects immune checkpoint inhibitor (ICI) efficacy. However, there is a paucity of clinical data evaluating the direct impact of endogenous GC on the efficacy for cancer patients with immune checkpoint blockade. Methods We first compared the endogenous circulating GC levels in healthy individuals and patients with cancer. We next retrospectively reviewed patients with advanced cancer with PD-1/PD-L1 inhibitor alone or combination therapy in a single center. The effects of baseline circulating GC levels on objective response rate (ORR), durable clinical benefit (DCB), progression-free survival (PFS), and overall survival (OS) were analyzed. The association of the endogenous GC levels with circulating lymphocytes, cytokines levels, and neutrophil to lymphocyte ratio, and tumor infiltrating immune cells, were systematically analyzed. Results The endogenous GC levels in advanced cancer patients were higher than those in early-stage cancer patients as well as healthy people. In the advanced cancer cohort with immune checkpoint blockade (n=130), patients with high baseline endogenous GC levels (n=80) had a significantly reduced ORR (10.0% vs 40.0%; p<0.0001) and DCB (35.0% vs 73.5%, p=0.001) compared to those with low endogenous GC levels (n=50). The increased GC levels was significantly associated with reduced PFS (HR 2.023; p=0.0008) and OS (HR 2.809; p=0.0005). Moreover, statistically significant differences regarding PFS, and OS were also detected after propensity score matching. In a multivariable model, the endogenous GC was identified as an independent indicator for predicting PFS (HR 1.779; p=0.012) and OS (HR 2.468; p=0.013). High endogenous GC levels were significantly associated with reduced lymphocytes (p=0.019), increased neutrophil to lymphocyte ratio (p=0.0009), and increased interleukin-6 levels (p=0.025). Patients with high levels of endogenous GC had low numbers of tumor infiltrating CD3+ (p=0.001), CD8+ T (p=0.059), and CD4+ T (p=0.002) cells, and the numbers of circulating PD-1+ NK cells (p=0.012), and the ratio of CD8+PD-1+ to CD4+PD-1+ (p=0.031) were higher in patients with high levels of endogenous GC compared to low levels of endogenous GC. Conclusion Baseline endogenous GC increase executes a comprehensive negative effect on immunosurveillance and response to immunotherapy in real-world cancer patients accompanied with cancer progression.
Collapse
Affiliation(s)
- Yu Cui
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Xinyue Han
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Hongtao Liu
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Yaping Guan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Beibei Yin
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Junjuan Xiao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Dongfeng Feng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Xuan Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Junwei Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Jinghua Chen
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Xingyu Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Weiwei Nie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Lin Ma
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Hairong Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Yan Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Baocheng Wang
- Department of Oncology, The 960th Hospital, The PEOPLE’s Liberation Army, Jinan, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
- *Correspondence: Jun Wang,
| |
Collapse
|
34
|
Zhou Z, Saluja AK, Houchen CW, Li M. Replication stress identifies novel molecular classification associated with treatment outcomes in pancreatic cancer. Pancreatology 2023; 23:82-89. [PMID: 36435734 DOI: 10.1016/j.pan.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Replication stress is a prominent hallmark of tumor cells, which is crucial for maintaining genomic integrity. However, it remains poorly understood whether replication stress can serve as a surrogate biomarker to indicate prognosis and treatment response of pancreatic cancer. METHODS Transcriptomic and clinical data were obtained from The Cancer Genome Atlas and literature. An integrated signature of 18 replication-stress associated genes (termed as REST18) was established using the cox proportional hazards regression analysis. Tumors were sorted into REST18-low and REST18-high groups. Survival analysis, gene set enrichment analysis and composition of immune cells were compared between these tumors. RESULTS Patients with REST18-high tumors showed worse prognoses than those with REST18-low tumors in the TCGA database and the finding is validated in an independent cohort of pancreatic cancer. Comparison of REST18 model and other molecular classifications showed that REST18-high tumors are positively correlated to basal-like or squamous phenotypes, which have higher metastasis potential. DNA repair pathway is enriched in the REST18-high tumors. Analysis of tumor immune microenvironment found that REST18-high tumors are characterized with "immune-cold" features. Univariate and multivariate analysis show that REST18 is an independent risk factor for overall survival and predicts outcomes of chemotherapy in pancreatic cancer. CONCLUSION REST18 is a novel biomarker to indicate prognosis and treatment response of chemotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anuj K Saluja
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
35
|
Zheng X, Gu H, Cao X, Pan B, Xiang H, Ju M, Xu S, Zheng M. Tislelizumab for cervical cancer: A retrospective study and analysis of correlative blood biomarkers. Front Immunol 2023; 14:1113369. [PMID: 36875089 PMCID: PMC9975598 DOI: 10.3389/fimmu.2023.1113369] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
Background Tislelizumab is an anti-programmed cell death 1 (PD-1) monoclonal antibody engineered to minimize binding to Fcγ receptors. It has been used to treat several solid tumors. However, its efficacy and toxicity, and the predictive and prognostic value of baseline hematological parameters in patients with recurrent or metastatic cervical cancer (R/M CC) receiving tislelizumab remain unclear. Methods We reviewed 115 patients treated for R/M CC with tislelizumab from March 2020 to June 2022 in our institute. The antitumor activity of tislelizumab was assessed using RECIST v1.1. Associations between the baseline hematological parameters and efficacy of tislelizumab in these patients were analyzed. Results With a median follow-up of 11.3 months (range, 2.2-28.7), the overall response rate was 39.1% (95% CI, 30.1-48.2) and the disease control rate was 77.4% (95% CI, 69.6-85.2). The median progression-free survival (PFS) was 19.6 months (95% CI, 10.7 to not reached). The median overall survival (OS) was not reached. Treatment-related adverse events (TRAEs) of any grade occurred in 81.7% of the patients and only 7.0% of the patients experienced grade 3 or 4 TRAEs. Univariate and multivariate regression analyses showed that the level of pretreatment serum C-reactive protein (CRP) was an independent risk factor for the response (complete or partial response) to tislelizumab and the PFS of R/M CC patients treated with tislelizumab (P = 0.0001 and P = 0.002, respectively). R/M CC patients with elevated baseline CRP levels had a short PFS (P = 0.0005). Additionally, the CRP-to-albumin ratio (CAR) was an independent risk factor for the PFS and OS of R/M CC patients treated with tislelizumab (P = 0.001 and P = 0.031, respectively). R/M CC patients with an elevated baseline CAR had short PFS and OS (P < 0.0001 and P = 0.0323, respectively). Conclusions Tislelizumab showed promising antitumor activity and tolerable toxicity in patients with R/M CC. The baseline serum CRP levels and CAR showed potential for predicting the efficacy of tislelizumab and the prognosis of R/M CC patients receiving tislelizumab.
Collapse
Affiliation(s)
- Xiaojing Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Haifeng Gu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xinping Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Baoyue Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Huiling Xiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Mingxiu Ju
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shijie Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Min Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Gynecology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Wu J, Zhou Z, Li J, Liu H, Zhang H, Zhang J, Huang W, He Y, Zhu S, Huo M, Liu M, Zhang C. CHD4 promotes acquired chemoresistance and tumor progression by activating the MEK/ERK axis. Drug Resist Updat 2023; 66:100913. [PMID: 36603431 DOI: 10.1016/j.drup.2022.100913] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022]
Abstract
AIMS Chemoresistance remains a major challenge in gastric cancer (GC). Chromodomain helicase DNA-binding protein 4 (CHD4) mediated chromatin remodeling plays critical roles in various tumor types, but its role in chemoresistance in GC remains uncharacterized. METHODS CHD4 expression was examined by immunohistochemistry and Western blotting. The role of CHD4 on cell proliferation and chemoresistance of GC was examined in vitro and in vivo. Immunoprecipitation and liquid chromatography-mass spectrometry were used to identify CHD4-binding proteins and a proximity ligation assay was used to explore protein-protein interaction. RESULTS Chemoresistance is associated with upregulation of CHD4 in the tumor tissues of GC patients. Overexpression of CHD4 increased chemoresistance and cell proliferation. Knockdown of CHD4 induced cell apoptosis and cell cycle arrest. CHD4 mediates the decrease of the intracellular concentration of cisplatin by inducing drug efflux. Additionally, CHD4 promotes the interaction between ERK1/2 and MEK1/2, resulting in continuous activation of MEK/ERK pathway. Knockdown of CHD4 in GC increased sensitivity to chemotherapy and suppressed tumor growth in a mouse xenograft model. CONCLUSIONS This study identifies CHD4 dominated multi-drug efflux as a promising therapeutic target for overcoming acquired chemoresistance in GC.
Collapse
Affiliation(s)
- Jing Wu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Department of Gastrointestinal Surgery of the First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2nd Road, Guangzhou 510080, Guangdong, China
| | - Zhijun Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Jin Li
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Huifang Liu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Huaqi Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Junchang Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Weibin Huang
- Department of Gastrointestinal Surgery of the First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2nd Road, Guangzhou 510080, Guangdong, China
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Department of Gastrointestinal Surgery of the First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2nd Road, Guangzhou 510080, Guangdong, China
| | - Shiyu Zhu
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China
| | - Mingyu Huo
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China.
| | - Mingyang Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Changhua Zhang
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China; Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, Guangdong, China.
| |
Collapse
|
37
|
Huang B, Lang X, Li X. The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front Oncol 2022; 12:1023177. [PMID: 36591515 PMCID: PMC9800921 DOI: 10.3389/fonc.2022.1023177] [Citation(s) in RCA: 173] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine involved in immune regulation. It can activate janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway. As one of the important signal transduction pathways in cells, JAK2/STAT3 signaling pathway plays a critical role in cell proliferation and differentiation by affecting the activation state of downstream effector molecules. The activation of JAK2/STAT3 signaling pathway is involved in tumorigenesis and development. It contributes to the formation of tumor inflammatory microenvironment and is closely related to the occurrence and development of many human tumors. This article focuses on the relationship between IL-6/JAK2/STAT3 signaling pathway and liver cancer, breast cancer, colorectal cancer, gastric cancer, lung cancer, pancreatic cancer and ovarian cancer, hoping to provide references for the research of cancer treatment targeting key molecules in IL-6/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Bei Huang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiaoling Lang
- Operational Management Office, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China,*Correspondence: Xiaoling Lang, ; Xihong Li,
| | - Xihong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China,Emergency Department, West China Second University Hospital, Sichuan University, Chengdu, China,*Correspondence: Xiaoling Lang, ; Xihong Li,
| |
Collapse
|
38
|
Liu Y, Li C, Lu Y, Liu C, Yang W. Tumor microenvironment-mediated immune tolerance in development and treatment of gastric cancer. Front Immunol 2022; 13:1016817. [PMID: 36341377 PMCID: PMC9630479 DOI: 10.3389/fimmu.2022.1016817] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
Tumor microenvironment is the general term for all non-cancer components and their metabolites in tumor tissue. These components include the extracellular matrix, fibroblasts, immune cells, and endothelial cells. In the early stages of tumors, the tumor microenvironment has a tumor suppressor function. As the tumor progresses, tumor immune tolerance is induced under the action of various factors, such that the tumor suppressor microenvironment is continuously transformed into a tumor-promoting microenvironment, which promotes tumor immune escape. Eventually, tumor cells manifest the characteristics of malignant proliferation, invasion, metastasis, and drug resistance. In recent years, stress effects of the extracellular matrix, metabolic and phenotypic changes of innate immune cells (such as neutrophils, mast cells), and adaptive immune cells in the tumor microenvironment have been revealed to mediate the emerging mechanisms of immune tolerance, providing us with a large number of emerging therapeutic targets to relieve tumor immune tolerance. Gastric cancer is one of the most common digestive tract malignancies worldwide, whose mortality rate remains high. According to latest guidelines, the first-line chemotherapy of advanced gastric cancer is the traditional platinum and fluorouracil therapy, while immunotherapy for gastric cancer is extremely limited, including only Human epidermal growth factor receptor 2 (HER-2) and programmed death ligand 1 (PD-L1) targeted drugs, whose benefits are limited. Clinical experiments confirmed that cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), vascular endothelial growth factor receptor (VEGFR) and other targeted drugs alone or in combination with other drugs have limited efficacy in patients with advanced gastric cancer, far less than in lung cancer, colon cancer, and other tumors. The failure of immunotherapy is mainly related to the induction of immune tolerance in the tumor microenvironment of gastric cancer. Therefore, solving the immune tolerance of tumors is key to the success of gastric cancer immunotherapy. In this study, we summarize the latest mechanisms of various components of the tumor microenvironment in gastric cancer for inducing immune tolerance and promoting the formation of the malignant phenotype of gastric cancer, as well as the research progress of targeting the tumor microenvironment to overcome immune tolerance in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Changfeng Li, ; Wei Yang,
| | - Yaoping Lu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
- *Correspondence: Changfeng Li, ; Wei Yang,
| |
Collapse
|
39
|
Qu L, Jin J, Lou J, Qian C, Lin J, Xu A, Liu B, Zhang M, Tao H, Yu W. The nuclear transportation of PD-L1 and the function in tumor immunity and progression. Cancer Immunol Immunother 2022; 71:2313-2323. [DOI: 10.1007/s00262-022-03176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/15/2022] [Indexed: 12/08/2022]
|
40
|
Wang Q, Guo F, Jin Y, Ma Y. Applications of human organoids in the personalized treatment for digestive diseases. Signal Transduct Target Ther 2022; 7:336. [PMID: 36167824 PMCID: PMC9513303 DOI: 10.1038/s41392-022-01194-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/09/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Digestive system diseases arise primarily through the interplay of genetic and environmental influences; there is an urgent need in elucidating the pathogenic mechanisms of these diseases and deploy personalized treatments. Traditional and long-established model systems rarely reproduce either tissue complexity or human physiology faithfully; these shortcomings underscore the need for better models. Organoids represent a promising research model, helping us gain a more profound understanding of the digestive organs; this model can also be used to provide patients with precise and individualized treatment and to build rapid in vitro test models for drug screening or gene/cell therapy, linking basic research with clinical treatment. Over the past few decades, the use of organoids has led to an advanced understanding of the composition of each digestive organ and has facilitated disease modeling, chemotherapy dose prediction, CRISPR-Cas9 genetic intervention, high-throughput drug screening, and identification of SARS-CoV-2 targets, pathogenic infection. However, the existing organoids of the digestive system mainly include the epithelial system. In order to reveal the pathogenic mechanism of digestive diseases, it is necessary to establish a completer and more physiological organoid model. Combining organoids and advanced techniques to test individualized treatments of different formulations is a promising approach that requires further exploration. This review highlights the advancements in the field of organoid technology from the perspectives of disease modeling and personalized therapy.
Collapse
Affiliation(s)
- Qinying Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fanying Guo
- School of Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yutao Jin
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Feng Z, Li K, Qin K, Liang J, Shi M, Ma Y, Zhao S, Liang H, Han D, Shen B, Peng C, Chen H, Jiang L. The LINC00623/NAT10 signaling axis promotes pancreatic cancer progression by remodeling ac4C modification of mRNA. J Hematol Oncol 2022; 15:112. [PMID: 35978332 PMCID: PMC9387035 DOI: 10.1186/s13045-022-01338-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although a substantial increase in the survival of patients with other cancers has been observed in recent decades, pancreatic ductal adenocarcinoma (PDAC) remains one of the deadliest diseases. No effective screening approach exists. METHODS Differential exosomal long noncoding RNAs (lncRNAs) isolated from the serum of patients with PDAC and healthy individuals were profiled to screen for potential markers in liquid biopsies. The functions of LINC00623 in PDAC cell proliferation, migration and invasion were confirmed through in vivo and in vitro assays. RNA pulldown, RNA immunoprecipitation (RIP) and coimmunoprecipitation (Co-IP) assays and rescue experiments were performed to explore the molecular mechanisms of the LINC00623/NAT10 signaling axis in PDAC progression. RESULTS A novel lncRNA, LINC00623, was identified, and its diagnostic value was confirmed, as it could discriminate patients with PDAC from patients with benign pancreatic neoplasms and healthy individuals. Moreover, LINC00623 was shown to promote the tumorigenicity and migratory capacity of PDAC cells in vitro and in vivo. Mechanistically, LINC00623 bound to N-acetyltransferase 10 (NAT10) and blocked its ubiquitination-dependent degradation by recruiting the deubiquitinase USP39. As a key regulator of N4-acetylcytidine (ac4C) modification of mRNA, NAT10 was demonstrated to maintain the stability of oncogenic mRNAs and promote their translation efficiency through ac4C modification. CONCLUSIONS Our data revealed the role of LINC00623/NAT10 signaling axis in PDAC progression, showing that it is a potential biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Zengyu Feng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
- Department of General Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Kexian Li
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Kai Qin
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Juyong Liang
- Otolaryngology and Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Minmin Shi
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yang Ma
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Shiwei Zhao
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Huaiyu Liang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Dongni Han
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, People's Republic of China
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Chenghong Peng
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China.
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China.
| | - Lingxi Jiang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China.
- Shanghai Key Laboratory of Translational Research for Pancreatic Neoplasms, Shanghai, People's Republic of China.
| |
Collapse
|
42
|
Yu Y. Multi-target combinatory strategy to overcome tumor immune escape. Front Med 2022; 16:208-215. [PMID: 35377102 DOI: 10.1007/s11684-022-0922-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/31/2021] [Indexed: 12/19/2022]
Abstract
Immune therapy has become the fourth approach after surgery, chemotherapy, and radiotherapy in cancer treatment. Many immune checkpoints were identified in the last decade since ipilimumab, which is the first immune checkpoint inhibitor to cytotoxic T-lymphocyte associated protein 4, had been approved by the US Food and Drug Administration (FDA) for the treatment of unresectable or metastatic melanoma in 2011. The use of several antibody drugs that target PD1/PD-L1 for various cancer treatments has been approved by the FDA. However, fewer people are benefitting from immune checkpoint inhibitor treatment in solid cancers. Approximately 80% of patients do not respond appropriately because of primary or acquired therapeutic resistance. Along with the characterization of more immune checkpoints, the combinatory treatment of multiimmune checkpoint inhibitors becomes a new option when monotherapy could not receive a good response. In this work, the author focuses on the combination therapy of multiple immune checkpoints (does not include targeted therapy of oncogenes or chemotherapy), introduces the current progression of multiple immune checkpoints and their related inhibitors, and discusses the advantages of combination therapy, as well as the risk of immune-related adverse events.
Collapse
Affiliation(s)
- Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
43
|
LI XM, YUAN DY, LIU YH, ZHU L, QIN HK, YANG YB, LI Y, YAN F, WANG YJ. Panax notoginseng saponins prevent colitis-associated colorectal cancer via inhibition IDO1 mediated immune regulation. Chin J Nat Med 2022; 20:258-269. [DOI: 10.1016/s1875-5364(22)60179-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Indexed: 12/11/2022]
|
44
|
Blockade of PD-L1/PD-1 signaling promotes osteo-/odontogenic differentiation through Ras activation. Int J Oral Sci 2022; 14:18. [PMID: 35365595 PMCID: PMC8976080 DOI: 10.1038/s41368-022-00168-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/17/2022] [Accepted: 03/08/2022] [Indexed: 12/23/2022] Open
Abstract
The programmed cell death ligand 1 (PD-L1) and its receptor programmed cell death 1 (PD-1) deliver inhibitory signals to regulate immunological tolerance during immune-mediated diseases. However, the role of PD-1 signaling and its blockade effect on human dental pulp stem cells (hDPSCs) differentiation into the osteo-/odontogenic lineage remain unknown. We show here that PD-L1 expression, but not PD-1, is downregulated during osteo-/odontogenic differentiation of hDPSCs. Importantly, PD-L1/PD-1 signaling has been shown to negatively regulate the osteo-/odontogenic differentiation of hDPSCs. Mechanistically, depletion of either PD-L1 or PD-1 expression increased ERK and AKT phosphorylation levels through the upregulation of Ras enzyme activity, which plays a pivotal role during hDPSCs osteo-/odontogenic differentiation. Treatment with nivolumab (a human anti-PD-1 monoclonal antibody), which targets PD-1 to prevent PD-L1 binding, successfully enhanced osteo-/odontogenic differentiation of hDPSCs through enhanced Ras activity-mediated phosphorylation of ERK and AKT. Our findings underscore that downregulation of PD-L1 expression accompanies during osteo-/odontogenic differentiation, and hDPSCs-intrinsic PD-1 signaling inhibits osteo-/odontogenic differentiation. These findings provide a significant basis that PD-1 blockade could be effective immunotherapeutic strategies in hDPSCs-mediated dental pulp regeneration.
Collapse
|
45
|
Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y, Xia Y. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol 2022; 15:24. [PMID: 35279217 PMCID: PMC8917703 DOI: 10.1186/s13045-022-01242-2] [Citation(s) in RCA: 246] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint molecules are promising anticancer targets, among which therapeutic antibodies targeting the PD-1/PD-L1 pathway have been widely applied to cancer treatment in clinical practice and have great potential. However, this treatment is greatly limited by its low response rates in certain cancers, lack of known biomarkers, immune-related toxicity, innate and acquired drug resistance, etc. Overcoming these limitations would significantly expand the anticancer applications of PD-1/PD-L1 blockade and improve the response rate and survival time of cancer patients. In the present review, we first illustrate the biological mechanisms of the PD-1/PD-L1 immune checkpoints and their role in the healthy immune system as well as in the tumor microenvironment (TME). The PD-1/PD-L1 pathway inhibits the anticancer effect of T cells in the TME, which in turn regulates the expression levels of PD-1 and PD-L1 through multiple mechanisms. Several strategies have been proposed to solve the limitations of anti-PD-1/PD-L1 treatment, including combination therapy with other standard treatments, such as chemotherapy, radiotherapy, targeted therapy, anti-angiogenic therapy, other immunotherapies and even diet control. Downregulation of PD-L1 expression in the TME via pharmacological or gene regulation methods improves the efficacy of anti-PD-1/PD-L1 treatment. Surprisingly, recent preclinical studies have shown that upregulation of PD-L1 in the TME also improves the response and efficacy of immune checkpoint blockade. Immunotherapy is a promising anticancer strategy that provides novel insight into clinical applications. This review aims to guide the development of more effective and less toxic anti-PD-1/PD-L1 immunotherapies.
Collapse
Affiliation(s)
- Mengling Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qianrui Huang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yao Xie
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, 610041, China
| | - Hongbo Ma
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Xia
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, 610041, China.
| |
Collapse
|
46
|
Yi K, Cui X, Liu X, Wang Y, Zhao J, Yang S, Xu C, Yang E, Xiao M, Hong B, Fang C, Kang C, Tan Y, Wang Q. PTRF/Cavin-1 as a Novel RNA-Binding Protein Expedites the NF-κB/PD-L1 Axis by Stabilizing lncRNA NEAT1, Contributing to Tumorigenesis and Immune Evasion in Glioblastoma. Front Immunol 2022; 12:802795. [PMID: 35069587 PMCID: PMC8778801 DOI: 10.3389/fimmu.2021.802795] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/14/2021] [Indexed: 12/23/2022] Open
Abstract
Background Immunotherapy, especially checkpoint inhibitors targeting PD-1 or PD-L1, has revolutionized cancer therapy. However, PD-1/PD-L1 inhibitors have not been investigated thoroughly in glioblastoma (GBM). Studies have shown that polymerase 1 and transcript release factor (PTRF/Cavin-1) has an immune-suppressive function in GBM. Thus, the relationship between PTRF and PD-L1 and their role in immune suppression requires further investigation in GBM. Methods We used public databases and bioinformatics analysis to investigate the relationship between PTRF and PD-L1. We next confirmed the predicted relationship between PTRF and PD-L1 in primary GBM cell lines by using different experimental approaches. RIP-Seq, RIP, ChIP, and qRT-PCR were conducted to explore the molecular mechanism of PTRF in immunosuppression. Results We found that PTRF stabilizes lncRNA NEAT1 to induce NF-κB and PD-L1 and promotes immune evasion in GBM. PTRF was found to correlate with immunosuppression in the public GBM databases. PTRF increased the level of PD-L1 in primary cell lines from GBM patients. We carried out RIP-Seq of GBM cells and found that PTRF interacts with lncRNA NEAT1 and stabilizes its mRNA. PTRF also promoted the activity of NF-κB by suppressing UBXN1 expression via NEAT1 and enhanced the transcription of PD-L1 through NF-κB activation. Finally, PTRF promoted immune evasion in GBM cells by regulating PD-1 binding and PD-L1 mediated T cell cytotoxicity. Conclusions In summary, our study identified the PTRF-NEAT1-PD-L1 axis as a novel immune therapeutic target in GBM.
Collapse
Affiliation(s)
- Kaikai Yi
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neuro-Oncology and Neurosurgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoteng Cui
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunfei Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Jixing Zhao
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Shixue Yang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Can Xu
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Eryan Yang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Menglin Xiao
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China.,Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Baoding, China
| | - Biao Hong
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Chuan Fang
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China.,Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Baoding, China
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanli Tan
- Key Laboratory of Precise Diagnosis and Treatment of Glioma in Hebei Province, Baoding, China.,Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China.,Department of Pathology, Hebei University School of Basic Medical Sciences, Baoding, China
| | - Qixue Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Key Laboratory of Post-Neuro Injury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| |
Collapse
|
47
|
Kim Y, Park S, Lee J, Jang J, Jung J, Koh JH, Choi CS, Wolfe RR, Kim IY. Essential Amino Acid-Enriched Diet Alleviates Dexamethasone-Induced Loss of Muscle Mass and Function through Stimulation of Myofibrillar Protein Synthesis and Improves Glucose Metabolism in Mice. Metabolites 2022; 12:metabo12010084. [PMID: 35050206 PMCID: PMC8778336 DOI: 10.3390/metabo12010084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 01/18/2023] Open
Abstract
Dexamethasone (DEX) induces dysregulation of protein turnover, leading to muscle atrophy and impairment of glucose metabolism. Positive protein balance, i.e., rate of protein synthesis exceeding rate of protein degradation, can be induced by dietary essential amino acids (EAAs). In this study, we investigated the roles of an EAA-enriched diet in the regulation of muscle proteostasis and its impact on glucose metabolism in the DEX-induced muscle atrophy model. Mice were fed normal chow or EAA-enriched chow and were given daily injections of DEX over 10 days. We determined muscle mass and functions using treadmill running and ladder climbing exercises, protein kinetics using the D2O labeling method, molecular signaling using immunoblot analysis, and glucose metabolism using a U-13C6 glucose tracer during oral glucose tolerance test (OGTT). The EAA-enriched diet increased muscle mass, strength, and myofibrillar protein synthesis rate, concurrent with improved glucose metabolism (i.e., reduced plasma insulin concentrations and increased insulin sensitivity) during the OGTT. The U-13C6 glucose tracing revealed that the EAA-enriched diet increased glucose uptake and subsequent glycolytic flux. In sum, our results demonstrate a vital role for the EAA-enriched diet in alleviating the DEX-induced muscle atrophy through stimulation of myofibrillar proteins synthesis, which was associated with improved glucose metabolism.
Collapse
Affiliation(s)
- Yeongmin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea; (Y.K.); (J.L.); (J.J.)
| | - Sanghee Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Korea; (S.P.); (J.-H.K.); (C.S.C.)
| | - Jinseok Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea; (Y.K.); (J.L.); (J.J.)
| | - Jiwoong Jang
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
- Gil Medical Center, Department of Internal Medicine, Gachon University, Incheon 21565, Korea
| | - Jiyeon Jung
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea; (Y.K.); (J.L.); (J.J.)
| | - Jin-Ho Koh
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Korea; (S.P.); (J.-H.K.); (C.S.C.)
| | - Cheol Soo Choi
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Korea; (S.P.); (J.-H.K.); (C.S.C.)
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
- Gil Medical Center, Department of Internal Medicine, Gachon University, Incheon 21565, Korea
| | - Robert R. Wolfe
- The Center for Translational Research in Aging and Longevity, Department of Geriatrics, Donald W. Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Korea; (S.P.); (J.-H.K.); (C.S.C.)
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
- Correspondence: ; Tel.: +82-32-899-6685
| |
Collapse
|
48
|
Yu Y. Repurposing glucocorticoids as adjuvant reagents for immune checkpoint inhibitors in solid cancers. Cancer Biol Med 2021; 18:j.issn.2095-3941.2021.0491. [PMID: 34697935 PMCID: PMC8610151 DOI: 10.20892/j.issn.2095-3941.2021.0491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/13/2021] [Indexed: 11/11/2022] Open
Affiliation(s)
- Yingyan Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
49
|
Glucocorticoid and PD-1 Cross-Talk: Does the Immune System Become Confused? Cells 2021; 10:cells10092333. [PMID: 34571982 PMCID: PMC8468592 DOI: 10.3390/cells10092333] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) and its ligands, PD-L1/2, control T cell activation and tolerance. While PD-1 expression is induced upon T cell receptor (TCR) activation or cytokine signaling, PD-L1 is expressed on B cells, antigen presenting cells, and on non-immune tissues, including cancer cells. Importantly, PD-L1 binding inhibits T cell activation. Therefore, the modulation of PD-1/PD-L1 expression on immune cells, both circulating or in a tumor microenvironment and/or on the tumor cell surface, is one mechanism of cancer immune evasion. Therapies that target PD-1/PD-L1, blocking the T cell-cancer cell interaction, have been successful in patients with various types of cancer. Glucocorticoids (GCs) are often administered to manage the side effects of chemo- or immuno-therapy, exerting a wide range of immunosuppressive and anti-inflammatory effects. However, GCs may also have tumor-promoting effects, interfering with therapy. In this review, we examine GC signaling and how it intersects with PD-1/PD-L1 pathways, including a discussion on the potential for GC- and PD-1/PD-L1-targeted therapies to "confuse" the immune system, leading to a cancer cell advantage that counteracts anti-cancer immunotherapy. Therefore, combination therapies should be utilized with an awareness of the potential for opposing effects on the immune system.
Collapse
|